Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Mater Chem B ; 12(24): 5957-5973, 2024 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-38808630

RESUMO

Doxorubicin (DOX), a chemotherapy drug, has demonstrated limited efficacy against glioblastoma, an aggressive brain tumor with resistance attributed to the blood-brain barrier (BBB). This study aims to overcome this challenge by proposing the targeted delivery of magnetic Janus nanoparticles (MJNPs) functionalized with folic acid ligands, fluorescent dye, and doxorubicin (DOX/MJNPs-FLA). The properties of these nanoparticles were comprehensively evaluated using bio-physiochemical techniques such as Fourier transform infrared (FTIR) spectroscopy, dynamic light scattering (DLS), zeta potential analysis, high-resolution transmission electron microscopy (HR-TEM), vibrating sample magnetometry (VSM), fluorescence microscopy, MTT assay, hemolysis assay, and liver enzyme level evaluation. Dual-controlled DOX release was investigated under different pH and temperature conditions. Additionally, the impact of DOX/MJNPs-FLA on apoptosis induction in tumor cells, body weight, and survival time of cancerous animals was assessed. The targeted delivery system was assessed using C6 and OLN-93 cell lines as representatives of cancerous and healthy cell lines, respectively, alongside Wistar rat tumor-bearing models. Results from Prussian blue staining and confocal microscopy tests demonstrated the effective targeted internalization of MJNPs-FLA by glioblastoma cells. Additionally, we investigated the biodistribution of the nanoparticles utilizing fluorescence imaging techniques. This enabled us to track the distribution pattern of MJNPs-FLA in vivo, shedding light on their movement and accumulation within the biological system. Furthermore, the combination of chemotherapy and magnetic hyperthermia exhibited enhanced efficacy in inducing apoptosis, as evidenced by the increase of the pro-apoptotic Bax gene and a decrease in the anti-apoptotic Bcl-2 gene. Remarkably, this combination treatment did not cause any hepatotoxicity. This study highlights the potential of DOX/MJNPs-FLA as carriers for therapeutic and diagnostic agents in the context of theranostic applications for the treatment of brain malignancies. Additionally, it demonstrates the promising performance of DOX/MJNPs-FLA in combination treatment through passive and active targeting.


Assuntos
Doxorrubicina , Ácido Fólico , Glioblastoma , Ratos Wistar , Animais , Doxorrubicina/química , Doxorrubicina/farmacologia , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Ratos , Ácido Fólico/química , Concentração de Íons de Hidrogênio , Antibióticos Antineoplásicos/farmacologia , Antibióticos Antineoplásicos/química , Nanomedicina Teranóstica , Temperatura , Ligantes , Sistemas de Liberação de Medicamentos , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Nanopartículas de Magnetita/química , Apoptose/efeitos dos fármacos , Portadores de Fármacos/química , Masculino , Humanos , Liberação Controlada de Fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos
2.
Cancer Cell Int ; 24(1): 160, 2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38715021

RESUMO

In recent times, the intersection of nanotechnology and biomedical research has given rise to nanobiomedicine, a captivating realm that holds immense promise for revolutionizing diagnostic and therapeutic approaches in the field of cancer. This innovative fusion of biology, medicine, and nanotechnology aims to create diagnostic and therapeutic agents with enhanced safety and efficacy, particularly in the realm of theranostics for various malignancies. Diverse inorganic, organic, and hybrid organic-inorganic nanoparticles, each possessing unique properties, have been introduced into this domain. This review seeks to highlight the latest strides in targeted glioblastoma therapy by focusing on the application of inorganic smart nanoparticles. Beyond exploring the general role of nanotechnology in medical applications, this review delves into groundbreaking strategies for glioblastoma treatment, showcasing the potential of smart nanoparticles through in vitro studies, in vivo investigations, and ongoing clinical trials.

3.
Int J Pharm ; 653: 123888, 2024 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-38342325

RESUMO

The goal of this work was to examine the heat-sensitizing effects of Janus-coated magnetic nanoparticles (JMNPs) as a vehicle for 5-fluorouracil (5-Fu) and Quercetin (Qu) in C6 and OLN-93 cell lines. The cellular uptake of nanoparticles was evaluated using Prussian blue staining and ICP-OES after monolayer culturing of C6 (rat brain cancer cell) and OLN-93 (normal rat brain cell) cells. The cells were treated with free 5-Fu, Qu, and MJNPs loaded with Qu/5-Fu for 24 h, followed by magnetic hyperthermia under an alternating magnetic field (AMF) at a temperature of 43 °C. Using the MTT test and Flow cytometry, the C6 and OLN-93 cells were investigated after being subjected to hyperthermia with and without magnetic nanoparticles. The results of Prussian blue staining confirmed the potential of MJNPs as carriers that facilitate the uptake of drugs by cancer cells. The results showed that the combined application of Qu/5-Fu/MJNPs with hyperthermia significantly increased the amount of ROS production compared to interventions without MJNPs. The therapeutic results demonstrated that the combination of Qu/5-Fu/MJNPs with hyperthermia considerably enhanced the rate of apoptotic and necrotic cell death compared to that of interventions without MJNPs. Furthermore, MTT findings indicated that controlled exposure of Qu/5-Fu/MJNPs to AMF caused a synergistic effect. The advanced Janus magnetic nanoparticles in this study can be proposed as a promising dual drug carrier (Qu/5-Fu) and thermosensitizer platform for dual-modal synergistic cancer therapy.


Assuntos
Ferrocianetos , Hipertermia Induzida , Nanopartículas , Polietilenoglicóis , Polietilenoimina , Ratos , Animais , Nanogéis , Preparações de Ação Retardada , Hipertermia Induzida/métodos , Fluoruracila , Linhagem Celular Tumoral , Quercetina/farmacologia
4.
Sci Rep ; 13(1): 22358, 2023 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-38102193

RESUMO

Malignant neoplasms are one of the main causes of death, especially in children, on a global scale, despite strenuous efforts made at advancing both diagnostic and therapeutic modalities. In this regard, a new nanocarrier Vincristine (VCR)-loaded Pluronic f127 polymer-coated magnetic nanoparticles conjugated with folic acid and transferrin (PMNP-VCR-FA-TF) were synthesized and characterized by various methods. The cytotoxicity of these nanoparticles was evaluated in vitro and ex vivo conditions. The in vitro anti-tumor effect of the nanoparticles was evaluated by colony formation assay (CFA) and reactive oxygen species (ROS) in Y79 cell line. The results showed that nanoparticles with two ligands conferred greater toxicity toward Y79 cancer cells than ARPE19 normal cells. Under an alternating magnetic field (AMF), these nanoparticles demonstrated a high specific absorption rate. The CFA and ROS results indicated that the AMF in combination with PMNP-VCR-FA-TF conferred the highest cytotoxicity toward Y79 cells compared with other groups (P < 0.05). PMNP-VCR-FA-TF could play an important role in converting externally applied radiofrequency energy into heat in cancer cells. The present study confirmed that dual targeting chemo-hyperthermia using PMNP-VCR-FA-TF was significantly more effective than hyperthermia or chemotherapy alone, providing a promising platform for precision drug delivery as an essential component in the chemotherapy of retinoblastoma.


Assuntos
Hipertermia Induzida , Nanopartículas de Magnetita , Nanopartículas , Neoplasias da Retina , Retinoblastoma , Criança , Humanos , Retinoblastoma/tratamento farmacológico , Espécies Reativas de Oxigênio , Ácido Fólico , Transferrina , Vincristina/farmacologia , Vincristina/uso terapêutico , Neoplasias da Retina/tratamento farmacológico , Linhagem Celular Tumoral
5.
Toxicol Appl Pharmacol ; 481: 116754, 2023 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-37956929

RESUMO

Glioblastoma multiforme (GBM) is one of the most vascular among solid tumors, and despite the use of multimodal therapies, the survival of these patients is poor. In order to target angiogenesis in GBM as a promising strategy, an antiangiogenic drug is required. This study was designed to evaluate the effects of sunitinib, a multityrosine kinase inhibitor with tumor proliferation and angiogenesis inhibitory properties, on GBM-bearing rats. Given the ineffective drug delivery to the brain due to the presence of the blood-brain barrier (BBB), intra-nasal (IN) drug delivery has recently been considered as a non-invasive method to bypass BBB. Therefore, in the current study, IN was used as an ideal method for the delivery of sunitinib to the brain, and the effects of this method were also compared to the OR administration of the sunitinib. GBM was induced in the brain of male Wistar rats, and they were randomly divided into 4 groups; IN-STB (sunitinib intranasal delivery), IN-sham (placebo intranasal delivery), OR-STB (sunitinib oral delivery) and OR-sham (placebo oral delivery). After the end of the treatment period, an MRI of animals' brains showed a reduction in tumor growth in the treatment groups. Immunohistochemistry revealed that sunitinib inhibits angiogenesis in GBM in both OR and IN delivery methods. Analysis of liver tissue and enzymes showed that IN delivery of sunitinib had less hepatotoxicity than the OR method. Overall, it was found that IN sunitinib delivery could be used as a potential non-hepatotoxic alternative for the treatment of GBM.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Animais , Humanos , Masculino , Ratos , Angiogênese , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Glioblastoma/tratamento farmacológico , Ratos Wistar , Sunitinibe/uso terapêutico
6.
Artigo em Inglês | MEDLINE | ID: mdl-37778031

RESUMO

A pivotal cause of death in the modern world, cancer is an insidious pathology that should be diagnosed at an early stage for successful treatment. Development of therapeutic interventions with minimal invasiveness and high efficacy that can discriminate between tumor and normal cells is of particular interest to the clinical science, as they can enhance patient survival. Nanoparticles are an invaluable asset that can be adopted for development of such diagnostic and therapeutic modalities, since they come in very small sizes with modifiable surface, are highly safe and stable, and can be synthesized in a controlled fashion. To date, different nanoparticles have been incorporated into numerous modalities such as tumor-targeted therapy, thermal therapy, chemotherapy, and radiotherapy. This review article seeks to deliver a brief account of recent advances in research and application of nanoparticles in hyperthermia-based cancer therapies. The most recent investigations are summarized to highlight the latest advances in the development of combined thermo-chemo-radiotherapy, along with the challenges associated with the application of nanoparticles in cancer therapy. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.


Assuntos
Hipertermia Induzida , Nanopartículas , Neoplasias , Humanos , Neoplasias/tratamento farmacológico , Nanomedicina , Nanopartículas/uso terapêutico , Quimiorradioterapia
7.
J Cancer Res Clin Oncol ; 149(10): 8087-8116, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37010586

RESUMO

Exosomes are nanosized extracellular vesicles secreted by nearly all viable cells following the fusing of multivesicular bodies and the plasma membrane and discharged into the encircling bodily fluids. Exosomes can transport cell-specific components from the source cell to the target cell. Given the enormous potential of exosomes as non-invasive diagnostic biomarkers and therapeutic nanovehicles. Lately, accumulated evidence has demonstrated that exosomes serve an important role in prognosis, diagnosis, and even treatment strategies. While several reviews have collective information on the biomedical application of exosomes, a comprehensive review incorporating updated and improved methodologies for beneficial applications of such vesicles in cancer theranostics is indispensable. In the current review, we first provided a comprehensive review of the introduction of exosomes, featuring their discovery, separation, characterization, function, biogenesis, secretion. The implications of exosomes as promising nanovehicles for drug and gene delivery, application of exosome inhibitors in the management of cancers, completed and ongoing clinical trials on the biological relevance of exosomes are then discussed in detail. As the field of exosome research grows, a better understanding of the subcellular parts and mechanisms involved in exosome secretion and targeting of specific cells will help figure out what their exact physiological functions are in the body.


Assuntos
Exossomos , Vesículas Extracelulares , Neoplasias , Humanos , Transporte Biológico , Exossomos/metabolismo , Vesículas Extracelulares/metabolismo , Neoplasias/diagnóstico , Neoplasias/terapia , Neoplasias/metabolismo , Medicina de Precisão , Ensaios Clínicos como Assunto
8.
Med Oncol ; 39(9): 122, 2022 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-35716197

RESUMO

The aim of this work was to assess the cytotoxicity, genotoxicity, and histopathological effects of Fe2O3@Au-FA NPs using in vitro and in vivo models. Cytotoxicity and cellular uptake of nanoparticles (NPs) by HUVECs were examined via 3-(4, 5-Dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay and inductively coupled plasma-mass-spectrometry (ICP-MS). This safe dose was then used for cytotoxicity assays, including total protein, total antioxidant capacity, lipid peroxidation, cell membrane integrity, reactive oxygen species, enzyme activity, and DNA damage. In the animal model, 32 Wistar rats were randomly categorized into 4 groups and received intraperitoneal injections of NPs. Blood samples for biochemical properties and histopathological changes were investigated. MTT results indicated 20 µg/ml as the safe dose for NPs. According to ICP-MS, treated cells showed significantly higher levels of the intracellular content of Fe (p < 0.001) and Au (p < 0.01) compared with the control group. In vitro tests did not show any significant cytotoxicity or genotoxicity at the safe dose of NPs. We found no significant elevation in intracellular γ-H2AX levels after treatment of HUVEC cells with Fe2O3@Au core-shell NPs (P > 0.05). As for the in vivo analysis, we observed no marked difference in serum biochemical parameters of rats treated with 50 mg/kg and 100 mg/kg doses of our NPs. Histopathological assessments indicated that liver, kidney, and testis tissues were not significantly affected at 50 mg/kg (liver), 50 mg/kg, and 100 mg/kg (kidney and testis) on NPs administration. These findings imply that the nanotoxicity of Fe2O3@Au-FA NPs in HUVECs and animals depends largely on the administrated dose. Our study suggests that Fe2O3@Au-FA NPs at a safe dose could be considered as new candidates in nanobiomedicine.


Assuntos
Ácido Fólico , Nanopartículas , Animais , Dano ao DNA , Compostos Férricos/química , Compostos Férricos/farmacologia , Ácido Fólico/química , Ácido Fólico/farmacologia , Marcadores Genéticos , Humanos , Masculino , Estresse Oxidativo , Ratos , Ratos Wistar
9.
Cell Biol Int ; 46(7): 1009-1020, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35322909

RESUMO

Almost a century after the devastating pandemic of the Spanish flu, humankind is facing the relatively comparable global outbreak of COVID-19. COVID-19 is an infectious disease caused by SARS-CoV-2 with an unprecedented transmission pattern. In the face of the recent repercussions of COVID-19, many have argued that the clinical experience with influenza through the last century may have tremendous implications in the containment of this newly emerged viral disease. During the last 2 years, from the emergence of COVID-19, tremendous advances have been made in diagnosing and treating coinfections. Several approved vaccines are available now for the primary prevention of COVID-19 and specific treatments exist to alleviate symptoms. The present review article aims to discuss the pathophysiology, diagnosis, and treatment of SARS-CoV-2 and influenza A virus coinfection while delivering a bioinformatics-based insight into this subject matter.


Assuntos
COVID-19 , Coinfecção , Influenza Pandêmica, 1918-1919 , Influenza Humana , Orthomyxoviridae , Coinfecção/diagnóstico , Coinfecção/epidemiologia , Biologia Computacional , História do Século XX , Humanos , Influenza Humana/epidemiologia , Influenza Humana/prevenção & controle , SARS-CoV-2
10.
Blood Purif ; 51(1): 1-14, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-33789273

RESUMO

Since early 2020, COVID-19 has wreaked havoc in many societies around the world. As of the present, the SARS-CoV-2-borne disease is propagating in almost all countries, affecting hundreds of thousands of people in an unprecedented way. As the name suggests, the novel coronavirus, widely known as SARS-CoV-2, is a new emerging human pathogen. A novel disease of relatively unknown origin, COVID-19 does not seem to be amenable to the currently available medicines since there is no specific cure for the disease. In the absence of any vaccine or effective antiviral medication, we have no tools at our disposal, but the method of quarantine, be it domestic or institutional, to hinder any further progression of this outbreak. However, there is a record of physicians in the past who practiced convalescent blood transfusion. To their awe, the method seemed to be useful. It is anticipated that these contemporary methods will outdo any other vaccination process in the time being, as blood transfusion is instead a cost-effective and time-friendly technique. Following a successful trial, this new approach of contemporary nature to a viral disease may serve as an emergency intervention to intercept infectious outbreaks and prevent an impending epidemic/pandemic. In this review, we document the most recent evidence regarding the efficiency of convalescent plasma and serum therapy on SARS, MERS, and particularly COVID-19, while discussing potential advantages and possible risks of such practice.


Assuntos
COVID-19/terapia , Pandemias , SARS-CoV-2 , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/uso terapêutico , COVID-19/epidemiologia , COVID-19/história , COVID-19/prevenção & controle , Ensaios Clínicos como Assunto , Convalescença , Infecções por Coronavirus/terapia , Previsões , História do Século XX , Humanos , Imunização Passiva/efeitos adversos , Imunização Passiva/ética , Imunização Passiva/história , Imunização Passiva/tendências , Influenza Humana/terapia , Plasma , Risco , SARS-CoV-2/imunologia , Soro , Síndrome Respiratória Aguda Grave/terapia , Soroterapia para COVID-19
11.
ACS Omega ; 6(31): 20192-20204, 2021 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-34395970

RESUMO

This study reports a new procedure for utilizing 5-fluorouracil (5-Fu)-loaded polycaprolactone (PCL)/chitosan-covered magnetite nanographene oxide (5-Fu/SPION/NGO@PCL-LMWC) as a platform for synergistic thermo-chemotherapy. In fact, superparamagnetic iron oxide nanoparticles/nanographene oxide (SPION/NGO) nanoparticles can be coated with copolymers PCL/chitosan to attain better colloidal stability in the biological environment. Nanoparticles were synthesized and characterized for their size, surface charge, X-ray patterns, polymer content, and in vitro heat-triggered release. In vitro cytotoxic effects of nanoparticles on CT-26 cells were assessed with an MTT assay and real-time polymerase chain reaction. In vivo tumor growth inhibition was evaluated on an allograft mouse model of CT-26 cells. Tumor-bearing mice were injected with 5-Fu-loaded nanoparticles intravenously, and then, the targeted delivery was amplified using a magnetic field and finally exposed to an alternating magnetic field (AMF) (40 A/m, 13.56 MHz), during which the tumor site temperature increased to 43 °C. By using an infrared camera, we managed to heat the nanoparticles up to a constant temperature between 42.5 and 43.5 °C, with a tolerance ±0.03 °C. Finally, in vitro results showed that 5-Fu-loaded nanoparticles combined with AMF hyperthermia significantly reduced the plating efficiency of the cells (P < 0.01) and increased the Bax/Bcl-2 ratio (1.42 times, P < 0.01) compared with those achieved with each one alone. Furthermore, in vivo results demonstrated that the treatment of 5-Fu-loaded nanoparticles combined with the AMF diminished the growth of CT-26 tumor cells and increased the life span of the tumor-bearing mice (P < 0.001) by thermal energy deposition compared to that of the free 5-Fu drug. Also, the high level of accumulation of the nanoparticles within the tumor site was easily monitored with magnetic resonance imaging. It was concluded that the multifunctional magnetic nanoparticles could be used as a promising nanocarrier platform for achieving concurrent goals, drug delivery, magnetic targeting, thermal-sensitizing, cell death induction, and real-time monitoring of response to treatment.

12.
Cell Mol Bioeng ; 14(4): 365-377, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34295445

RESUMO

INTRODUCTION: With an emphasis on the radioresistant nature of glioblastoma cells, the aim of the present study was to evaluate the radio-thermo-sensitizing effects of PCL-PEG-coated Superparamagnetic iron oxide nanoparticles (SPIONs) as a carrier of 5-iodo-2-deoxyuridine (IUdR) in monolayer culture of U87MG human glioma cell line. METHODS: Following monolayer culture of U87MG cells, nanoparticle uptake was assessed using Prussian blue staining and ICP-OES method. The U87MG cells were treated with an appropriate concentration of free IUdR and PCL-PEG-coated SPIONs (MNPs) loaded with IUdR (IUdR/MNPs) for 24 h, subjected to hyperthermia (water bath and alternating magnetic field (AMF)) at 43 °C, and exposed to X-ray (2 Gy, 6 MV). The combined effects of hyperthermia with or without magnetic nanoparticles on radiosensitivity of the U87MG cells were evaluated using colony formation assay (CFA) and Flowcytometry. RESULTS: Prussian blue staining and ICP-OES showed that the nanoparticles were able to enter the cells. The results also indicated that IUdR/MNPs combined with X-ray radiation and hyperthermia significantly decreased the colony formation ability of monolayer cells (1.11, 1.41 fold) and increased the percentage of apoptotic (2.47, 4.1 fold) and necrotic cells (12.28, 29.34 fold), when compared to IUdR combined with X-ray and hyperthermia or IUdR/MNPs + X-ray. MTT results revealed that the presence of IUdR/MNPs significantly increased the toxicity of AMF hyperthermia compared to the water bath method. CONCLUSIONS: Our study showed that SPIONs/PCL-PEG, as a carrier of IUdR, can enhance the cytotoxic effects of radiotherapy and hyperthermia and act as a radio-thermo-sensitizing agent. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1007/s12195-021-00675-y.

13.
Mol Pharmacol ; 100(2): 119-143, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33990406

RESUMO

Most commonly recognized as a catabolic pathway, autophagy is a perplexing mechanism through which a living cell can free itself of excess cytoplasmic components, i.e., organelles, by means of certain membranous vesicles or lysosomes filled with degrading enzymes. Upon exposure to external insult or internal stimuli, the cell might opt to activate such a pathway, through which it can gain control over the maintenance of intracellular components and thus sustain homeostasis by intercepting the formation of unnecessary structures or eliminating the already present dysfunctional or inutile organelles. Despite such appropriateness, autophagy might also be considered a frailty for the cell, as it has been said to have a rather complicated role in tumorigenesis. A merit in the early stages of tumor formation, autophagy appears to be salutary because of its tumor-suppressing effects. In fact, several investigations on tumorigenesis have reported diminished levels of autophagic activity in tumor cells, which might result in transition to malignancy. On the contrary, autophagy has been suggested to be a seemingly favorable mechanism to progressed malignancies, as it contributes to survival of such cells. Based on the recent literature, this mechanism might also be activated upon the entry of engineered nanomaterials inside a cell, supposedly protecting the host from foreign materials. Accordingly, there is a good chance that therapeutic interventions for modulating autophagy in malignant cells using nanoparticles may sensitize cancerous cells to certain treatment modalities, e.g., radiotherapy. In this review, we will discuss the signaling pathways involved in autophagy and the significance of the mechanism itself in apoptosis and tumorigenesis while shedding light on possible alterations in autophagy through engineered nanomaterials and their potential therapeutic applications in cancer. SIGNIFICANCE STATEMENT: Autophagy has been said to have a complicated role in tumorigenesis. In the early stages of tumor formation, autophagy appears to be salutary because of its tumor-suppressing effects. On the contrary, autophagy has been suggested to be a favorable mechanism to progressed malignancies. This mechanism might be affected upon the entry of nanomaterials inside a cell. Accordingly, therapeutic interventions for modulating autophagy using nanoparticles may sensitize cancerous cells to certain therapies.


Assuntos
Antineoplásicos/farmacologia , Neoplasias/patologia , Transdução de Sinais , Antineoplásicos/uso terapêutico , Apoptose , Autofagia/efeitos dos fármacos , Progressão da Doença , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Nanotecnologia , Estadiamento de Neoplasias , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Transdução de Sinais/efeitos dos fármacos
14.
Mater Sci Eng C Mater Biol Appl ; 124: 112043, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33947543

RESUMO

Normal tissue complication and development of radioresistance in cancer cells are known as the main challenges of ionizing radiation treatment. In the current study, we intended to induce selective radiosensitization in HT29 cancer cells by developing folic acid modified magnetic triblock copolymer nanoparticles as carrier of 5-Flourouracil (5-FU) which was further used in combination with hyperthermia. The aforementioned nanoparticles were synthesized and characterized by differential scanning calorimetric analysis (DSC), UV-visible spectroscopy, dynamic light scattering (DLS), zeta sizer, and transmission electron microscopy (TEM). These nanoparticles were also assessed to determine drug loading capacity (DLC %) and drug release profile. The cytotoxicity of nanoparticles was evaluated on two different cell lines: HUVEC and HT29. Furthermore, radiosensitivity induction of the nanoparticles with and without exposure of alternative magnetic field was investigated. MTT-based cytotoxicity assay demonstrated that the therapeutic ratio was enhanced in response to using 5-FU-loaded nanoparticles as compared to 5-FU. Various characterizations including gene expression study, measurement of reactive oxygen species (ROS) generation, Annexin V/PI staining, and clonogenic assay revealed that ionizing radiation in combination with hyperthermia in the presence of the synthesized nanoparticles led to maximal anti-cancer effects as compared to other single (P < 0.001) and combined treatments (P < 0.01). Our results suggested that combined treatment based on using folic acid modified magnetic copolymer nanoparticle as carrier of 5-FU accompanied with hyperthermia could be proposed as an efficient approach to enhance radiation effects in cancer cells.


Assuntos
Fluoruracila , Nanopartículas , Linhagem Celular Tumoral , Fluoruracila/farmacologia , Células HT29 , Humanos , Hipertermia , Fenômenos Magnéticos , Tolerância a Radiação
15.
Colloids Surf B Biointerfaces ; 203: 111731, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33831752

RESUMO

Reversible and remote cell manipulation with high spatiotemporal precision is now a highly attractive subject in various biological applications such as tissue engineering and cell-matrix interaction. Herein, photoresponsive poly(methyl methacrylate-co-hydroxy ethyl methacrylate-co-spiropyran ethyl acrylate) terpolymer (MHSP) was prepared using emulsion polymerization and the corresponding nanofibers (MHSP@NF) and film (MHSP@F) were prepared using electrospinning and drop-casting techniques, respectively. Structure of MHSP@NF with cylindrical cross-section and uniform diameter size of 169 nm were characterized by 1H-NMR and SEM analyses. Time-dependent UV-vis spectra of the prepared acrylic nanofibers and films demonstrated maximum forward photoisomerization after 3- and 8-min UV irradiation at 365 nm together with a 96° and 5° decrement in their surface water contact angles, respectively. High photoresponsivity of the nanofibers was attributed to their extensive surface area which exposes more spiropyran groups to UV light. MHSP@F and MHSP@NF with chemically-attached spiropyran groups demonstrated significant biocompatibility with negligible toxicity toward C6 glioma cancer cells up to 5 days. However, MH/SP@NF with doped SPOH exhibited a sudden decrease in cell viability relating to the migration and leakage of SPOH molecules. Photoreversible cell adhesion results showed a dramatic and switchable C6 cells attachment/detachment upon alternating UV and visible lights irradiations for MHSP@NF sample, while this was not observed for the similar film. These indicate potentiality of MHSP@NF as a promising substrate for dynamic switching of biomolecules and cell sheet engineering.


Assuntos
Glioma , Nanofibras , Benzopiranos , Humanos , Indóis , Nitrocompostos
16.
Appl Microbiol Biotechnol ; 105(7): 2615-2624, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33710356

RESUMO

A most discussed topic of the new decade, COVID-19 is an infectious disease caused by the recently discovered SARS-CoV-2. With an exceedingly high transmission rate, COVID-19 has affected almost all the countries in the world. Absent any vaccine or specific treatment, the humanity is left with nothing but the legacy method of quarantine. However, quarantine can only be effective when combined with early diagnosis of suspected cases. With their high sensitivity and unmatched specificity, biosensors have become an area of interest for development of novel diagnostic methods. Compared to the more traditional diagnostics, nanobiotechnology introduces biosensors as different diagnostics with greater versatility in application. Today, a growing number of analytes are being accurately identified by these nanoscopic sensing machines. Several reports of validated application with real samples further strengthen this idea. As of recent, there has been a rise in the number of studies on portable biosensors. Despite the slow progression, certain devices with embedded biosensors have managed to be of diagnostic value in several countries. The perceptible increase in development of mobile platforms has revolutionized the healthcare delivery system in the new millennium. The present article reviews the most recent advancements in development of diagnostic nanobiosensors and their application in the clinical fields. KEY POINTS: • There is no specific treatment for highly transmissible SARS-CoV-2. • Early diagnosis is critical for control of pandemic. • Highly sensitive/specific nanobiosensors are emerging assets against COVID-19.


Assuntos
Técnicas Biossensoriais/métodos , COVID-19/diagnóstico , Diagnóstico Precoce , SARS-CoV-2 , Técnicas Biossensoriais/instrumentação , Humanos , Técnicas de Diagnóstico Molecular , Nanotecnologia , Técnicas de Amplificação de Ácido Nucleico , SARS-CoV-2/isolamento & purificação
17.
J Neurooncol ; 152(3): 419-428, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33713248

RESUMO

INTRODUCTION: Hyperthermia therapy (HT) is a recognized treatment modality, that can sensitize tumors to the effects of radiotherapy (RT) and chemotherapy by heating up tumor cells to 40-45 °C. The advantages of noninvasive inductive magnetic hyperthermia (MH) over RT or chemotherapy in the treatment of recurrent/progressive glioma have been confirmed by several clinical trials. Thus, here we have conducted a systematic review to provide a concise, albeit brief, account of the currently available literature regarding this topic. METHODS: Five databases, PubMed/Medline, Embace, Ovid, WOS, and Scopus, were investigated to identify clinical studies comparing overall survival (OS) following RT/chemotherapy versus RT/chemotherapy + MH. RESULTS: Eleven articles were selected for this systematic review, including reports on 227 glioma patients who met the study inclusion criteria. The papers included in this review comprised nine pilot clinical trials, one non-randomized clinical trial, and one retrospective investigation. As the clinical trials suggested, MH improved OS in primary glioblastoma (GBM), however, in the case of recurrent glioblastoma, no significant change in OS was reported. All 11 studies ascertained that no major side effects were observed during MH therapy. CONCLUSION: Our systematic review indicates that MH therapy as an adjuvant for RT could result in improved survival, compared to the therapeutic outcomes achieved with RT alone in GBM, especially by intratumoral injection of magnetic nanoparticles. However, heterogeneity in the methodology of the most well-known studies, and differences in the study design may significantly limit the extent to which conclusions can be drawn. Thus, further investigations are required to shed more light on the efficacy of MH therapy as an adjuvant treatment modality in GBM.


Assuntos
Glioblastoma , Hipertermia Induzida , Glioblastoma/terapia , Glioma/terapia , Humanos , Fenômenos Magnéticos , Recidiva Local de Neoplasia/terapia , Estudos Retrospectivos
18.
Nanomedicine ; 31: 102319, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33068745

RESUMO

The goal of this study was to develop a new method based on Oncothermia with concomitant use of the temozolomide (TMZ)-loaded magnetic nanoparticles conjugated with folic acid (TMZ/MNPs-FA) and alternative magnetic field (AMF) and evaluate its efficacy in the treatment of C6 glioma in rats. TMZ/MNPs-FA were prepared and evaluated for their size, surface charge, magnetic saturation, hemolysis and in vitro AMF-triggered release. The glioma rat models were treated with free TMZ, MNPs-FA and TMZ/MNPs-FA in the presence or absence of AMF (43 °C). The results confirmed that a combinatorial therapy consisting of AFM hyperthermia and thermosensitive TMZ/MNPs-FA could significantly suppress tumor growth, increase survival rate and promote apoptosis (P < 0.0001). Therefore, this treatment strategy may be a powerful modality for treatment of cancer, as the thermal and mechanical effects of magnetic nanoparticles exposed to AMF can increase the therapeutic efficacy of conventional chemotherapy.


Assuntos
Glioma/tratamento farmacológico , Campos Magnéticos , Nanopartículas de Magnetita/química , Animais , Ácido Fólico/uso terapêutico , Ratos , Temozolomida/uso terapêutico
19.
Nanomedicine (Lond) ; 15(23): 2255-2270, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32975155

RESUMO

Aim: In this study, the effects of ionizing radiation and 5-fluorouracil (5-FU)-loaded triblock copolymer-coated magnetic nanoparticles (NPs) on the induction of apoptosis in HT-29 and HCT-116 were investigated. Materials & methods: The percentage of apoptotic cells and alteration of the expression of apoptotic-related proteins were evaluated in treated cells by flow cytometry and western blot analysis, respectively. Results: Combination treatment with 5-FU and radiation had a stronger effect on decreasing Bcl-2 expression and increasing expression of Bax, cleaved caspase-9, cleaved caspase-3, cleaved PARP compared with each treatment alone. Conclusion: The combination of radiation and triblock copolymer-coated magnetic NPs as 5-FU drug carriers works by triggering apoptosis to improve in vitro treatment efficacy. Additional study may present the NPs as an effective approach for the treatment of colon cancer.


Assuntos
Neoplasias do Colo , Nanopartículas de Magnetita , Nanopartículas , Antimetabólitos Antineoplásicos/uso terapêutico , Apoptose , Linhagem Celular Tumoral , Neoplasias do Colo/tratamento farmacológico , Fluoruracila/uso terapêutico , Humanos , Raios X
20.
J Cell Physiol ; 235(12): 8873-8924, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32452539

RESUMO

Months after the outbreak of a new flu-like disease in China, the entire world is now in a state of caution. The subsequent less-anticipated propagation of the novel coronavirus disease, formally known as COVID-19, not only made it to headlines by an overwhelmingly high transmission rate and fatality reports, but also raised an alarm for the medical community all around the globe. Since the causative agent, SARS-CoV-2, is a recently discovered species, there is no specific medicine for downright treatment of the infection. This has led to an unprecedented societal fear of the newly born disease, adding a psychological aspect to the physical manifestation of the virus. Herein, the COVID-19 structure, epidemiology, pathogenesis, etiology, diagnosis, and therapy have been reviewed.


Assuntos
Betacoronavirus/patogenicidade , Técnicas de Laboratório Clínico , Infecções por Coronavirus/diagnóstico , Infecções por Coronavirus/terapia , Sistema Imunitário/virologia , Pneumonia Viral/diagnóstico , Pneumonia Viral/terapia , Betacoronavirus/imunologia , COVID-19 , Teste para COVID-19 , China/epidemiologia , Infecções por Coronavirus/virologia , Humanos , Pandemias , Pneumonia Viral/virologia , Sistema Respiratório/virologia , SARS-CoV-2
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...